Language selection

Search

Patent 2168950 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2168950
(54) English Title: METHODS FOR EX VIVO THERAPY USING PEPTIDE-LOADED ANTIGEN PRESENTING CELLS FOR THE ACTIVATION OF CTL
(54) French Title: METHODES POUR THERAPIE EX VIVO, UTILISANT DES ANTIGENES CHARGES DE PEPTIDES, AVEC PRESENTATION DE CELLULES POUR L'ACTIVATION DES CTL
Status: Dead
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 5/0783 (2010.01)
  • A61K 35/14 (2006.01)
  • A61K 39/00 (2006.01)
(72) Inventors :
  • CELIS, ESTEBAN (United States of America)
  • KUBO, RALPH (United States of America)
  • SERRA, HORACIO (United States of America)
  • TSAI, VAN (United States of America)
  • WENTWORTH, PEGGY (United States of America)
(73) Owners :
  • EPIMMUNE INC. (United States of America)
(71) Applicants :
(74) Agent: FETHERSTONHAUGH & CO.
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 1994-08-01
(87) Open to Public Inspection: 1995-02-16
Examination requested: 2001-11-14
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US1994/008672
(87) International Publication Number: WO1995/004817
(85) National Entry: 1996-02-06

(30) Application Priority Data:
Application No. Country/Territory Date
08/103,401 United States of America 1993-08-06

Abstracts

English Abstract






Methods for activating cytotoxic T lymphocytes (CTL) in vitro are presented in conjunction with methods for using the activated
CTL for therapy in vivo. Additionally, a method for killing specific CTL in vivo is presented using antigen presenting cells which were
modified in vitro.


French Abstract

L'invention concerne des procédés qui permettent d'activer des CTL (lymphocytes T cytotoxiques) in vitro, en conjonction avec des procédés qui permettent d'utiliser ces CTL activés en thérapie in vivo. En outre, l'invention concerne un procédé permettant de tuer in vivo des CTL spécifiques à l'aide de cellules présentatrices d'antigènes modifiées in vitro.

Claims

Note: Claims are shown in the official language in which they were submitted.


39
WHAT IS CLAIMED IS:
1. A method for activating cytotoxic T cells in
vitro comprising:
dissociating bound peptides from class I MHC molecules
on antigen presenting cells, using a mild acid treatment;
associating desired immunogenic peptides with the
class I MHC molecules on the antigen presenting cells; and
incubating the antigen presenting cells with the
cytotoxic T cells in the presence of a growth factor, thereby
producing activated cytotoxic T cells.

2. The method of claim 1, wherein the step of
dissociating bound peptides is carried out by incubating the
antigen presenting cells in a glycine or citrate-phosphate
buffer solution at pH 3.

3. The method of claim 1, wherein the step of
associating desired immunogenic peptides with the MHC molecules
is carried out by incubating the antigen presenting cells with
about 10 to 50µg/ml immunogenic peptide.

4. The method of claim 1, wherein the step of
incubating the antigen presenting cells with the cytotoxic T
cells for about 7 to about 10 days.

5. The method of claim 1 wherein the antigen
presenting cells are peripheral blood mononuclear cells isolated
from a patient.

6. The method of claim 5 wherein the peripheral
blood mononuclear cells are SAC-I activated.

7. The method of claim 1 wherein the incubating step
includes a growth factor.

8. The method of claim 8 wherein the growth factor
is IL-7 and said growth factor is added at day 0 and day 7.


9. The method of claim 7 wherein the growth factor
is IL-2 and said growth factor is added after day 7.

10. The method of claim 1, further comprising:
contacting the activated cytotoxic T cells with an acceptable
carrier, thereby forming a pharmaceutical composition; and
administering the pharmaceutical composition to a patient.

11. The method of claim 10, further comprising
separating the activated cytotoxic T cells from the antigen
presenting cells.

12. The method of claim 10 wherein the cytotoxic T
cells are useful in the treatment of cancer, AIDS, hepatitis,
bacterial infection, fungal infection, malaria or tuberculosis.

13. A method of specifically killing target cells in
a human patient, comprising:
obtaining a fluid sample containing cytotoxic T cells
from the patient;
contacting the cytotoxic T cells with antigen
presenting cells comprising class I MHC molecules having
selected immunogenic peptides associated therewith, thereby
producing activated cytotoxic T cells;
contacting the activated cytotoxic T cells with an
acceptable carrier, thereby forming a pharmaceutical
composition; and
administering the pharmaceutical composition to a
patient.

14. The method of claim 13, further comprising the
step of dissociating bound peptides from the antigen presenting
cells by incubating the antigen presenting cells in a glycine or
citrate-phosphate buffer solution at pH 3.

15. The method of claim 12, further comprising the
step of associating desired immunogenic peptides with the MHC
molecules on the antigen presenting cells by incubating the

41
antigen presenting cells with about 10 to 50µg/ml immunogenic
peptide.

16. The method of claim 12 wherein the antigen
presenting cells are peripheral blood mononuclear cells isolated
from a patient.

17. The method of claim 12, wherein the step of
incubating the antigen presenting cells with the cytotoxic T
cells for about 7 to about 10 days.

Description

Note: Descriptions are shown in the official language in which they were submitted.


WO95/~17 ~ 6 ~ 9 ~ ~ PCT~S94/08672




METHODS FOR EX VIVO THERAPY USING PEPTIDE-LOADED ANTIGEN
PRESENTING CELLS FOR THE ACTIVATION OF CTL

BACKGROUND OF THE INV~:N110N
The present invention relates to compositions and
methods for preventing or treating a number of pathological
states such as viral diseases and cancer through ex vivo
therapy. In particular, it provides methods for inducing
cytotoxic T lymphocytes (CTL) using antigen presenting cells
(APC) with a peptide of choice bound to selected major
histocompatibility complex (MHC) molecules.
Cytotoxic T cells, or CD8 cells as they are also
known, represent the main line of defense against viral
infections. CTLS specifically recognize and kill cells which
are infected by a virus. The T cell receptors on the surface
of CTLS cannot recognize foreign antigens directly. In
contrast to antibodies, antigen must first be presented to the
T cell receptors for activation to occur.
The presentation of antigen to T cells is
accomplished by the major histocompatibility complex (MHC)
molecules. The major histocompatibility complex (MHC) refers
to a large genetic locus encoding an extensive family of
glycoproteins which play an important role in the immune
response. The MHC genes, which are also referred to as the
HLA (human leukocyte antigen) complex, are located on
chromosome 6 in humans. The molecules encoded by MHC genes
are present on cell surfaces and are largely responsible for
recognition of tissue transplants as "non-self".
MHC molecules are classified as either Class I,
Class II or class III molecules. Class II MHC molecules are
expressed primarily on cells involved in initiating and
sustaining immune responses, such as T lymphocytes, B
lymphocytes, macrophages, etc. Class II MHC molecules are
recognized by helper T lymphocytes and induce proliferation of
helper T lymphocytes and amplification of the immune response
r

WO95/04817 ~ PCT~S94/08672
21689~ 0 2
to the particular immunogenic peptide that is displayed.
Class I MHC molecules are expressed on almost all nucleated
cells and are recognized by CTLs. T cells that serve mainly
as helper cells express CD4 and are primarily restricted to
Class II molecules, whereas CD8-expressing cells, represented
by cytotoxic effector cells, interact with Class I molecules.
The CTL recognizes the antigen in the form of a
peptide fragment bound to the MHC class I molecules rather -
than the intact foreign antigen itself. The antigen must
normally be endogenously synthesized by the cell, and a
portion of the protein antigen is degraded into small peptide
fragments in the cytoplasm. Some of these small peptides
translocate into a pre-Golgi compartment and interact with
class I heavy chains to facilitate proper folding and
association with the subunit B2 microglobulin. The
peptide-MHC class I complex is then routed to the cell surface
for expression and potential recognition by specific CTLs.
Investigations of the crystal structure of the human MHC class
I molecule, HLA-A2.1, indicate that a peptide binding groove
is created by the folding of the ~1 and ~2 domains of the
class I heavy chain (Bjorkman et al., Nature, 329:506 ( 1987).
For many years, immunologists have hoped to raise
specific cytotoxic cells targeting viruses, retroviruses and
cancer cells. One possible approach is to immunize a healthy
individual, isolate the CTLs from this individual, and inject
these cells into the diseased person. This experimental
protocol seems to work in inbred mouse strains, but it has not
been successfully tried in humans. For this approach to work
the MHC haplotype of the donor must be identical to that of
the recipient. This is important because the CTLs of the
recipient can only interact with peptides bound to one of the
three to six Class I molecules present in the individual.
Second, CTLs react violently with all Class I molecules which
are different from those expressed in the individual from whom
the CD8 cells are obtained, regardless of what peptides the
Class I molecules contain. This reactivity is the underlying
cause of the immune rejection of transplanted organs.

WO9S/~17 ~ 6 ~ g 5 0 PCT~sg4/08672

Because it is difficult to find two unrelated
persons with exactly the same Class I molecules, some
therapeutic approaches take the non-specific approach of
"boosting" existing CD8 cells by incubating them in vitro with
IL-2, a growth factor for T cells. However, this protocol
(known as LAK cell therapy or TIL [tumor infiltrating
lymphocytes] therapy) will only allow the expansion of those
CTLs which are already activated. As the immune system is -
always active for one reason or another, most of the IL-2
stimulated cells will be irrelevant for the purpose of
combatting the disease. In fact, it has not been documented
that this type of therapy activates an~ cells with the desired
specificity. The benefits of LAK cell therapy are ambiguous
at best, and the side effects are often severe. (Greenberg,
P. 1991. Adoptive T cell therapy of tumors: Mechanisms
operative in the recognition and elimination of tumor cells.
Advances in Immunology 49:281. Melief, C. 1992. Tumor
eradication by adoptive transfer of cytotoxic T lymphocytes.
Adv. Cancer Research 58:14. 34. Riddell, S., K. Watanabe, J.
Goodrich, C. Li, M. Agha, P. Greenberg. 1992. Restoration of
viral immunity in immunodeficient humans by the adoptive
transfer of T cell clones. Science 257:238. ).
The preferred approach for the treatment of such
diseases as cancer, AIDS, hepatitis and other infectious
disease would be to activate only those CTLs recognizing
diseased cells. While various procedures have been applied in
these diseases, few if any successful attempts using cytotoxic
T cells have been reported. Ex vivo activation of CTLs would
be the preferable means of treating the types of disease noted
above. However, no reliable procedures have been available to
specifically activate CTLs associated with these diseases.
The present invention addresses these and other problems.

SUMMARY OF THE INVENTION
This invention is directed to methods of activating
cytotoxic T cells (CD8 cells) in vitro or in vivo. The
methods of activating CD8 cells comprise: dissociating bound
peptides from class I MHC molecules on antigen presenting

wo 9~817 ~ ~ 6 8 9 ~ o PCT~S94/08672




cells using a mild acid treatment; associating selected
immunogenic peptides with the class I MHC molecule on the
antigen presenting cell; and incubating the antigen presenting
cells with the cytotoxic T cells, thereby producing activated
cytotoxic T cells. The methods of the present invention are
capable of generating empty MHC class I molecules on antigen
presenting cells and in turn inducing CTL and affecting
killing of class I matched cells.
The antigen presenting cells having empty MHC class
I molecules on their surface are capable of inducing cytotoxic
T cells which are useful in the treatment of chronic
infectious diseases and cancer. Specifically, this invention
provides methods of producing empty MHC class I molecules on
antigen presenting cells, loading those empty MHC class I
molecules with selected immunogenic peptides, activating
cytotoxic T cells which are specific for killing specific
antigen targets. This invention has broad therapeutic
application in the treatment of cancers, certain immune
diseases and viral diseases. As such the method may further
comprise: separating activated CTLs from the antigen
presenting cells having the empty MHC class I molecule on its
surface; suspending the activated CTLs in an acceptable
carrier or excipient as a pharmaceutical composition; and
administering the pharmaceutical composition to a patient
having the disease.

~168~50
WO95/~17 ~CT~S94/08672

BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the effects of ~2microglobulin and
exogenous 941.01 (HBC 18-27) peptide on MHC class I molecules
from acid stripped and loaded PHA blasts
Figure 2 shows CTL induction using GC43 A2.1
responders and autologous acid-stripped PBMCs or PHA blasts
loaded with the 777.03 (HBs 20-28); 924.07 (HBC 18-27); 927.32
(HBp 61-69) peptide pool.
Figure 3 shows CTL induction using X351 or X355 A2.1
responders and autologous acid stripped PBMCs or PHA blasts as
stimulators after loading with the 1044.04 (PAP 135-143);
1044.05 (PSA 166-175) 1044.06 (PSA 118-128) peptide pool.
Figure 4 shows CTL induction using GC49 A2.1
responders and Autologous Acid stripped PHA blasts as
stimulators after loading with 939.03 (PSA 49-57) peptide.
Figure 5 shows CTL induction using GC66 Al
responders and autologous acid stripped PBMCs as stimulators
after loading of peptide 958.01 (MAGE 1:161-169).
Figure 6 shows CTL induction using GC 30, HLA Al
responders and autologous cold temperature incubated SAC-I
activated PBMC~ as stimulators after loading with 1044.07
MAGE-3 (161-169) peptides.
Figure 7 shows a comparison of different methods to
load peptides onto SAC-I activated PBMCs as APCs. A pool of
MAGE-3 HLA Al binding peptides (1044.07:161-167 and 1044.01:8-
17) were tested with donor GC 164. 7 A-acid strip; 7 B- cold
temperature incubation; 7 C- room temperature, no
preincubation or acid strip with 4 hour peptide loading only;
7 p- room temperature, no acid stripping with addition of
soluble peptide to the culture.
Figure 8 shows the result of the CTL induction assay
using selected MAGE peptides.
Figure 9 shows the result of the CTL induction assay
using selected HIV peptides.
Figure 10 shows the result of the CTL induction
assay using selected HCV peptides.
Figure 11 shows the retuls of the CTL induction
assay using selected HBV peptides.

WO95/~17- PCT~S94/08672
~68~9~0
DESCRIPTION OF THE PREFERRED EMBODIMENTS
The term "peptide" is used interchangeably with
"oligopeptide" in the present specification to designate a
series of residues, typically L-amino acids, connected one to
the other typically by peptide bonds between the alpha-amino
and carbonyl groups of adjacent amino acids.
An "immunogenic peptide" is a peptide which
comprises an allele-specific motif such that the peptide will
bind the MHC allele and be capable of inducing a CTL response.
Thus, immunogenic peptides are capable of binding to an
appropriate class I MHC molecule and inducing a cytotoxic T
cell response against the antigen from which the immunogenic
peptide is derived.
The term "residue" refers to an amino acid or amino
acid mimetic incorporated in an oligopeptide by an amide bond
or amide bond mimetic.
The present invention relates to methods of
enhancing the immune response to various diseases using ex
vivo therapy. The general approach of the invention comprises
isolation of peripheral blood mononuclear cells (PBMCs) from a
patient, loading a desired immunogenic peptide into the
binding pockets of MHC class I molecules on the surface of
antigen presenting cells (APCs), incubating the APCs with
precursor CTLs in the sample to induce proliferation of CTLs
recognizing the peptide, and using the CTLs to identify
antigenic epitopes and by expanding their numbers introduce
the activated CTLs into the patient.
The procedures of the present invention depend in
part upon the determination of epitopes recognized by CTLs
capable of eliminating target infected cells. One approach to
identification of these epitopes is the identification of
allele-specific peptide motifs associated with a particular
disease for human Class I MHC allele subtypes. The MHC class
I antigens are encoded by the HLA-A, B, and C loci. HLA-A and
B antigens are expressed at the cell surface at approximately
equal densities, whereas the expression of HLA-C is
significantly lower (perhaps as much as 10-fold lower). Each
of these loci have a number of alleles. A large number of

WO95/~17 ~ 8 ~ 5 0 PCT~S94/08672




cells with defined MHC molecules, particularly MHC Class I
molecules, are known and readily available. These cells can
be used to identify particular allele specific motifs
associated with target diseases.
The allele-specific motifs are then used to define T
cell epitopes from any desired antigen, particularly those
associated with human viral diseases or cancers, for which the
amino acid sequence of the potential antigen targets is known.
This general approach is described in detail in copending and
commonly assigned applications U.S.S.N. 07/926,666 and
U.S.S.N. 08/027,146, which are incorporated herein by
reference.
Potential epitopes on a number of target proteins
can be identified in this manner. Examples of suitable
antigens include prostate specific antigen (PSA), hepatitis B
core, surface and polymerase antigens (HBVc, HBVs, HBVp),
hepatitis C antigens, Epstein-Barr virus antigens, melanoma
antigens (e.g., MAGE-1), human immunodeficiency virus (HIV)
antigens, human papilloma virus (HPV) antigens,
cytomegalovirus (CMV), herpes simplex virus (HSV), and other
oncogene products (c-Erb B2, CEA, p 53-breast/ovary).
These approaches typically involve isolation of
peptides from a particular MHC molecule and sequencing the
peptides to determine the relevant motif. Buus et al.,
Science, 242:1065 (1988) first described a method for acid
elution of bound peptides from MHC. Subsequently, Rammensee
and his coworkers (Falk et al., Nature, 351:290 (1991)
developed an approach to characterize naturally processed
peptides bound to class I molecules. Other investigators have
successfully achieved direct amino acid sequencing of the more
abundant peptides in various HPLC fractions by conventional
automated sequencing of peptides eluted from B type class I
molecules (Jardetzky, et al., Nature, 353:326 (1991) and of
the A2.1 type by mass spectrometry (Hunt, et al., Science,
225:1261 (1992). A review of the characterization of
naturally processed peptides found on MHC Class I molecules is
presented by Rotzschke and Falk (Rotzschke and Falk, Immunol.
Today, 12:447 (1991).

WO95/~17 ~1 6 ~ ~ 5 0 PCT~S94/08672




Definition of motifs specific for different class I
alleles allows the identification of potential peptide
epitopes from an antigenic protein whose amino acid sequence
is known. Typically, identification of potential peptide
epitopes is initially carried out using a computer to scan the
amino acid sequence of a desired antigen for the presence of
motifs. The epitopic sequences are then synthesized. The
capacity to bind MHC Class molecules is measured in a variety
of different ways using, for example, purified class I
molecules and radioiodinated peptides and/or cells expressing
empty class I molecules by, for instance, immunofluorescent
staining and flow microfluorimetry, peptide-dependent class I
assembly assays, and inhibition of CTL recognition by peptide
competition. Other alternatives described in the literature
include inhibition of antigen presentation (Sette, et al., J.
Immunol ., 141:3893 (1991), in vitro assembly assays (Townsend,
et al., Cell, 62:285 (1990), and FACS based assays using
mutated ells, such as RMA.S (Melief, et al., Eur . J. Immunol .,
21:2963 [1991]).
Next, peptides that test positive in the MHC class I
binding assay are assayed for the ability of the peptides to
induce specific primary or secondary CTL responses in vitro.
For instance, antigen-presenting cells that have been
incubated with a peptide can be assayed for the ability to
induce CTL responses in responder cell populations. For
secondary responses, antigen-presenting cells can be normal
cells such as peripheral blood mononuclear cells or dendritic
cells (Inaba, et al., J. Exp. Ned., 166:182 (1987); Boog, Eur.
J. Immunol., 18:219 [1988]).
Alternatively, mutant mammalian cell lines that are
deficient in their ability to load class I molecules with
internally processed peptides, such as the mouse cell lines
RMA-S (Karre, et al.. Nature, 319:675 (1986~; Ljunggren, et
al., Eur . J. Immunol ., 21:2963-2970 (1991)), and the human
somatic T cell hybridoma, T-2 (Cerundolo, et al., Nature,
345:449-452 (1990)) and which have been transfected with the
appropriate human class I genes are conveniently used, when
peptide is added to them, to test for the capacity of the

WO95/~17 ~ 6 ~ 9 ~ O PCT~S94/08672




peptide to induce in vitro primary CTL responses. These empty
MH~ cells are preferable for inducing a primary response since
the density of MHC-peptide complexes on the surface of the
antigen presenting cell will be greater. Other eukaryotic
cell lines which could be used include various insect cell
lines such as mosquito larvae (ATCC cell lines CCL 125, 126,
1660, 1591, 6585, 6586), silkworm (ATTC CRL 8851), armyworm
(ATCC CRL 1711), moth (ATCC CCL 80) and Drosophila cell lines
such as a Schneider cell line that have been transfected with
the appropriate human class I MHC allele encoding genes and
the human B2 microglobulin genes.
Once the appropriate epitope is determined,
immunogenic peptides comprising the motif required for MHC
binding and the epitope recognized by the CTL are synthesized.
15 The immunogenic peptides can be prepared synthetically, or by
recombinant DNA technology or isolated from natural sources
such as whole viruses or tumors. One of skill will recognize
that the immunogenic peptides can be a variety of lengths,
either in their neutral (uncharged) forms or in forms which
are salts, and either free of modifications such as
glycosylation, side chain oxidation, or phosphorylation or
containing these modifications, subject to the condition that
the modification not destroy the biological activity of the
polypeptides as herein described.
Desirably, the peptide will be as small as possible
while still maintaining substantially all of the biological
activity of the large peptide. When possible, it may be
desirable to optimize peptides of the invention to a length of
9 or 10 amino acid residues, commensurate in size with
endogenously processed viral peptides or tumor cell peptides
that are bound to MHC class I molecules on the cell surface.
Peptides having the desired activity may be modified
as necessary to provide certain desired attributes, e.g.,
improved pharmacological characteristics, while increasing or
at least retaining substantially all of the biological
activity of the unmodified peptide to bind the desired MHC
molecule and activate the appropriate T cell. For instance,
the peptides may be subject to various changes, such as

W095/~17 PCT~S94/08672
21 68950 lO

substitutions, either conservative or non-conservative, where
such changes might provide for certain advantages in their
use, such as improved MHC binding. By conservative
substitutions is meant replacing an amino acid residue with
another which is biologically and/or chemically similar, e.g.,
one hydrophobic residue for another, or one polar residue for
another. The substitutions include combinations such as Gly,
Ala; Val, Ile, Leu, Met; Asp, Glu; Asn, Gln; Ser, Thr; Lys,
Arg; and Phe, Tyr. The effect of single amino acid
substitutions may also be probed using D-amino acids. Such
modifications may be made using well known peptide synthesis
procedures.
The peptides of the invention can be prepared in a
wide variety of ways. Because of their relatively short size,
the peptides can be synthesized in solution or on a solid
support in accordance with conventional techniques. Various
automatic synthesizers are commercially available and can be
used in accordance with known protocols. See, for example,
Stewart and Young, sol id Phase Peptide Synthesis, 2d. ed.,
Pierce Chemical Co. (1984), supra.
Alternatively, recombinant DNA technology may be
employed wherein a nucleotide sequence which encodes an
immunogenic peptide of interest is inserted into an expression
vector, transformed or transfected into an appropriate host
cell and cultivated under conditions suitable for expression.
These procedures are generally known in the art, as described
generally in Sambrook et al., Molecular Cloning, A Laboratory
Manual, Cold Spring Harbor Press, Cold Spring Harbor, New York
(1982), which is incorporated herein by reference. Fusion
proteins which comprise one or more peptide sequences of the
invention can also be used to present the appropriate T cell
epitope.
The immunogenic peptides are then used to activate
CTL ex vivo. The ex vivo therapy methods of the present
invention and pharmaceutical compositions thereof are useful
for treatment of mammals, particularly humans, to treat and/or
prevent viral infection, immune disorders and cancer.
Examples of diseases which can be treated using the ex vivo

W095/~17 21~ ~ 9 ~ O PCT~S94/08672
11 '
therapy methods of the invention include prostate cancer,
hepatitis B, hepatitis C, AIDS, renal carcinoma, cervical
carcinoma, lymphoma, CMV, condyloma acuminatum breast and
ovarian cancer, colon, lung cancer and HSV.
For therapeutic use, therapy should begin at the
first sign of viral infection or the detection or surgical
removal of tumors or shortly after diagnosis in the case of
acute infection. This is followed by boosting levels of CTL
at least until symptoms are substantially abated and for a
period thereafter. In chronic infection, loading doses
followed by boosting doses may be required.
Treatment of an infected individual with the methods
of the invention may hasten resolution of the infection in
acutely infected individuals. For those individuals
susceptible (or predisposed) to developing chronic infection
the methods are useful for preventing the evolution from acute
to chronic infection. Where the susceptible individuals are
identified prior to or during infection, the compositions can
be targeted to them, minimizing the need for administration to
a larger population.
The methods of the present invention can also be
used for the treatment of chronic infection and to stimulate
the immune system to eliminate virus-infected cells in
carriers.
Ex vivo CTL responses to a particular pathogen
(infectious agent or tumor antigen) are induced by incubating
in tissue culture the patient's CTL precursor cells (CTLp)
together with a source of antigen-presenting cells (APC)
loaded with the appropriate immunogenic peptide. After an
appropriate incubation time (typically 3-12 weeks) in which
the CTLp are activated and mature and expand into effector
CTL, the cells are infused back into the patient, where they
will destroy their specific target cell (an infected cell or a
tumor cell). Infusion of the cells into the patient may
include a T cell growth factor such as interleukin 2 (IL-2).
In order to optimize the in vitro conditions for the
generation of specific cytotoxic T cells, the culture of
stimulator cells is maintained in an appropriate serum-free

WO95/~17 PCT~S94/08672
~168950
12
medium which may include one or more growth factors such as
IL-2, IL-4, IL-7 and IL-12.
Peripheral blood lymphocytes are conveniently
isolated following simple venipuncture or leukapheresis of
normal donors or patients and used as the responder cell
sources of CTLp. In one embodiment particularly for secondary
CTL responses, the appropriate APC are incubated with 10-100
~M of peptide in serum-free media for 4 hours under
appropriate culture conditions. The peptide-loaded APC are
then incubated with the responder cell populations in vitro
for 7 to 10 days under optimized culture conditions. For
primary CTL induction, APC expressing empty MHC would be used
to stimulate naive CTLp. In this case the CTL would be
stimulated more frequently (1-2 times).
Positive CTL activation can be determined by
assaying the cultures for the presence of CTLs that kill
radiolabeled target cells, both specific peptide-pulsed
targets as well as target cells expressing endogenously
processed form of the relevant virus or tumor antigen from
which the peptide sequence was derived.
Specificity and MHC restriction of the CTL of a
patient can be determined by a number of methods known in the
art. For instance, CTL restriction can be determined by
testing against different peptide loaded target cells
expressing human MHC class I alleles shared with the HLA
phonotype of the donor CTL. The peptides that test positive
in the MHC binding assays and give rise to specific CTL
responses are identified as immunogenic peptides.
As mentioned above, the induction of CTL in vitro
requires the specific recognition of peptides that are bound
to allele specific MHC class I molecules on APC. The number
of specific MHC/peptide complexes per APC determines the level
of stimulation of CTL, particularly during the primary immune
response. While small amounts of peptide/MHC complexes per
cell are sufficient to render a cell susceptible to lysis by
CTL, or to stimulate a secondary CTL response, the successful
activation of a CTLp during primary response requires a
significantly higher number of MHC/peptide complexes.

WO95/~17 ~1~ 8 9 ~ ~ PCT~S~/08672
13
Since mutant cell lines capable of expressing empty
MHC do not exist for every human MHC allele, it is
advantageous to use a technique to remove endogenous
MHC-associated peptides from the surface of APC, followed by
loading the resulting empty MHC molecules with the immunogenic
peptides of interest. The use of non-transformed
(non-tumorigenic), non-infected cells, and preferably,
autologous cells of patients as APC is desirable for the
design of CTL induction protocols directed towards development
of ex vivo CTL therapies. This present invention provides
novel methods generating empty class I MHC which can then be
loaded with an appropriate immunogenic peptide by stripping
the endogenous MHC-associated peptides from the surface of APC
or through cold temperature incubation (37 C~26 C)followed by
the loading of desired peptides.
A stable MHC class I molecule is a trimeric complex
formed of the following elements: 1) a peptide usually of 8 -
10 residues, 2) a transmembrane heavy polymorphic protein
chain which bears the peptide-binding site in its ~1 and ~2
domains, and 3) a non-covalently associated non-polymorphic
light chain, ~2microglobulin. Removing the bound peptides
and/or dissociating the ~2microglobulin from the complex
renders the MHC class I molecules nonfunctional and unstable,
resulting in rapid degradation at 37 C. Almost all MHC class
I molecules isolated from PBMCs have endogenous peptides bound
to them. Therefore, the first step to prepare APC for primary
CTL induction is to remove all endogenous peptides bound to
MHC class I molecules on the APC without causing degradation
or cell death before exogenous peptides can be added.
Two possible ways to generate free MHC class I
molecules include lowering the culture temperature from 37C
to 26C overnight to allow NHC class I without peptides to be
expressed and stripping the endogenous peptides from the cell
using a mild acid treatment. The mild acid treatment releases
previously bound peptides into the extracellular environment
allowing new exogenous peptides to bind to the empty class I
molecules. The overnight cold-temperature incubation at 26C
which may slow the cell's metabolic rate enables expression of

WO95/~17 PCT~S94/08672
~16895~
14
stable empty class I molecules which then bind exogenous
peptides efficiently. It is also likely that cells not
actively synthesizing MHC molecules (e.g., resting PBMC) would
not produce high amounts of empty surface MHC molecules by the
cold temperature procedure.
Extraction of the peptides is accomplished by harsh
acid stripping using trifluoroacetic acid, pH 2, or acid
denaturation of the immunoaffinity purified class I-peptide
complexes. These methods are not feasible for CTL induction,
since it is important to remove the endogenous peptides while
preserving APC viability and an optimal metabolic state which
is critical for antigen presentation. Mild acid solutions of
pH 3 such as glycine or citrate-phosphate buffers have been
used to identify endogenous peptides and to identify tumor
associated T cell epitopes (31. Storkus, W., H. Zeh, R.
Salter, and M. Lotze. 1993. Identification of I' cell
epitopes: Rapid isolation of class I-presented peptides from
viable cells by mild acid elution [submitted]). The treatment
is especially effective, in that only the MHC class I
molecules are destabilized (and associated peptides released),
while other surface antigens remain intact, including MHC
class II molecules. (16. Suguwara, S., T. Abo, and K.
Kumagai. 1987. A simple method to eliminate the antigenicity
of surface class I MHC molecules from the membrane of viable
cells by acid treatment at pH 3. J. Immunol. Meth. 100:83).
Most importantly, treatment of cells with the mild acid
solutions do not affect the cell's viability or metabolic
state. The mild acid treatment is rapid since the stripping
of the endogenous peptides occurs in two minutes at 4C and
the APC is functional after the appropriate peptides are
loaded. The techn; que is utilized herein to make peptide-
specific APCs for the generation of primary antigen-specific
CTL. The resulting APCs are efficient in inducing
peptide-specific CTL.
Typically in a primary response prior to incubation
of the APCs with the CTLp to be activated, an amount of
antigenic peptide is added to the APCs or stimulator cell
culture, of sufficient quantity to become loaded onto the

2168950
WO9~ 17 ~ PCT~S94/08672

human Class I molecules to be expressed on the surface of the
APCs. In the present invention, a sufficient amount of
peptide is an amount that will allow about 200 or more human
Class I MHC molecules loaded with peptide to be expressed on
the surface of each stimulator cell. Preferably, the
stimulator cells are incubated with 5-lOO~g/ml peptide.
Resting or precursor CTLs are then incubated in
culture with the appropriate APCs for a time period sufficient
to activate the CTLs. The CTLs are activated in an antigen-
specific manner. The ratio of precursor CTLs to APCs may varyfrom individual to individual and may further depend upon
variables such as the amenability of an individual's
lymphocytes to culturing conditions and the nature and
severity of the disease condition or other condition for which
the within-described treatment modality is used. Preferably,
however, the CTL:APC (i.e. responder to stimulator) ratio is
in the range of about 10:1 to 100:1. The CTL/APC culture may
be maintained for as long a time as is necessary to stimulate
a therapeutically useable or effective number of CTL.
Activated CTL may be effectively separated from the
APC using one of a variety of known methods. For example,
monoclonal antibodies specific for the APCs, for the peptides
loaded onto the stimulator cells, or for the CTL (or a segment
thereof) may be utilized to bind their appropriate
complementary ligand. Antibody-tagged cells may then be
extracted from the admixture via appropriate means, e.g., via
well-known immunoprecipitation or immunoassay methods.
Effective, cytotoxic amounts of the activated CTLs
can vary between in vitro and in vivo uses, as well as with
the amount and type of cells that are the ultimate target of
these killer cells. The amount will also vary depending on
the condition of the patient and should be determined via
consideration of all appropriate factors by the practitioner.
Preferably, however, about 1 X 106 to about 1 X 1012, more
preferably about 1 X 108 to about 1 X 1011, and even more
preferably, about 1 X 109 to about 1 X 101 activated CTLS are
utilized for adult humans, compared to about 5 X 106 - 5 X 107
cells used in mice.

WO951~17 ~ PCT~S94/08672
2168350 16

As discussed above, the activated CTLS may be
harvested from the cell culture prior to administration of the
cells to the individual being treated. It is important to
note, however, that unlike other present treatment modalities,
the present method uses a cell culture system that does not
contain transformed or tumor cells. Therefore, if complete
separation of antigen-presenting cells and activated CTLS is
not achieved, there is no inherent danger known to be
associated with the administration of a small number of
stimulator cells, whereas administration of mammalian tumor-
promoting cells may be extremely hazardous.
one embodiment of the present invention uses the APC
generated by the in vitro techniques of this application for
therapy against CTL in vivo. In this embodiment, the APC are
a patient's cells (e.g., the peripheral blood cells) which are
stripped of their natural antigenic peptides and loaded with a
peptide of choice which is conjugated to a toxin (e.g. ricin A
chain or pseudomonas toxin). The APCs are then re-introduced
into the patient, where they will be bound by the endogenous
CTLs that are specific for the antigenic peptide. The coupled
toxin will kill the activated CTL that are harmful i.e. those
which stimulate transplant rejection after it binds the APC.
Such directed CTL killing is broadly useful for treating
tissue-transplantation rejection and auto-immune disorders,
which are mediated through CTL. The treatment regime will
vary depending upon the specific disorder to be treated and
the judgement of the treating physician.
Methods of re-introducing cellular components are
known in the art and include procedures such as those
exemplified in U.S. Patent No. 4,844,893 to Honsik, et al. and
U.S. Patent No. 4,690,915 to Rosenberg, which are incorporated
herein by reference. For example, administration of activated
CTLs via intravenous infusion is appropriate.

The following examples are offered by way of
illustration, not by way of limitation.

W095/~17 216 8 9 5 0 PCT~S94/08672


ExamPle 1
Ex vivo induction of Cytotoxic T LYmphocytes (CTL)

Peripheral blood mononuclear cells (PBMC) are
isolated from an HLA-typed patient by either venipuncture or
leukapheresis (depending upon the initial amount of CTLp
required), and purified by gradient centrifugation using
Ficoll-Paque (Pharmacia). Typically, one can obtain one
million PBMC for every ml of peripheral blood, or
alternatively, a typical leukapheresis procedure can yield up
to a total of 1-10 X 101 PBMC.
The isolated and purified PBMC are co-cultured with
an appropriate number of APC expressing empty MHC molecules,
previously incubated ("pulsed") with an appropriate amount of
synthetic peptide (containing the HLA binding motif and the
sequence of the antigen in question). PBMC are usually
incubated at 1-3 X 106 cells/ml in culture medium such as
RPMI-1640 (with autologous serum or plasma) or the serum-free
medium AIM-V (Gibco).
APC are usually used at concentrations ranging from
lX104 to lX106 cells/ml, depending on the type of cell used.
Possible sources of APC include: autologous PBMCs, SAC-I
activated PBMCs, PHA blasts; autologous dendritic cells (DC)
which are isolated from PBMC and purified as described (Inaba,
et al., J. Exp. Ned., 166:182 (1987)); and mutant and
genetically engineered mammalian cells such as the mouse RMA-S
cell line or the human T2 cell line transfected with the
appropriate MHC genes that express "empty" HLA molecules which
are syngeneic to the patient's allelic HLA form). APC
containing empty HLA molecules are known to be potent inducers
of CTL responses, possibly because the peptide can associate
more readily with empty MHC molecules than with MHC molecules
which are occupied by other peptides (DeBruijn, et al., Eur.
J. Immunol., 21:2963-2970 (1991)).
The APC are gamma irradiated with an appropriate
dose (using, e.g., radioactive cesium or cobalt) to prevent
their proliferation and to facilitate the expansion of the
CTLp.

W095/~17 ~1 6 8 9 5 ~ PCT~S94/08672
18
The mixture cultures, containing PBMC, APC and
peptide are kept in an appropriate culture vessel such as
plastic T-flasks, gas-permeable plastic bags, or roller
bottles, at 37 centigrade in a humid air/C02 incubator.
After the activation phase of the culture, which usually
occurs during the first 3-5 days, the resulting effector CTL
can be further expanded, by the addition of recombinant growth
factors such as interleukin-2 (IL-2), interleukin-4 (IL-4), or
interleukin-7 (IL-7) to the cultures. An expansion culture
can be kept for an additional 5 to 12 days, depending on the
numbers of effector CTL required for a particular patient. In
addition, expansion cultures may be performed using hollow
fiber artificial capillary systems (Cellco), where larger
numbers of cells (up to lX1011) can be maintained. In order
to obtain the required cell numbers for treatment, it may be
necessary to restimulate the cultures 2-4 times with
irradiated, autologous, peptide pulsed adherent PBMCs.
Before the cells are infused into the patient, they
are tested for activity, viability, toxicity and sterility.
The cytotoxic activity of the resulting CTL can be determined
by a standard 5ICr-release assay (Biddison, W.E. 1991, Current
Protocols in Immunology, p7,17.1-7.17.5, Ed. J. Coligan et
al., J. Wiley and Sons, New York), using target cells that
express the appropriate HLA molecule, in the presence and
absence of the immunogenic peptide. Viability is determined
by the exclusion of trypan blue dye by live cells. Cells are
tested for the presence of endotoxin by conventional
t~chn;ques. Finally, the presence of bacterial or fungal
contamination is determined by appropriate microbiological
methods (chocolate agar, etc.). Once the cells pass all
quality control and safety tests, they are washed and placed
in the appropriate infusion solution (Ringer/glucose
lactate/human serum albumin) which may include a T-cell growth
factor such as IL-2 and infused intravenously into the
patient.

WO95/~17 ~1 6 8 9 S ~ PCT~S94/08672

19
Exam~le 2
PreParation of effective HLA allele-specific antiqen
~resentinq cells bY acid stri~Ping followed by peptide
loadinq.
This example demonstrates the use of cold
temperature incubation and acid stripping for generation of
empty MHC class I molecules to enable peptide loading method
to prepare effective HLA-allele-specific antigen presenting
cells (APC) for use in diagnostic or ex vivo therapy
applications. The APC in this example were used to sensitize
precursor cytotoxic T lymphocytes for the development of
antigen-specific cytotoxic cells. This was accomplished using
either staphylococcus aureus cowan I SAC I activated PBMC,
phytohemagglutinin (PHA) T-cell blasts or peripheral blood
mononuclear cells (PBMC) as APC in the HLA-A2.1 and HLA-A1
systems. The results are applicable to other APC and to the
other MHC alleles.

Culture Medium. PHA blasts and CTL inductions were done in
RPMI 1640 + Hepes + glutamine (Gibco) supplemented with 2 mM
L-glutamine (Irvine Scientific), 50 ~g/ml gentamicin (Gibco),
and 5% heat inactivated pooled human Type AB serum (Gemini
Bioproducts) [RPMI/5% HS]. EBV transformed lymphoblastoid
cell lines (LCL) were maintained in RPMI 1640 + Hepes +
glutamine (BioWhittaker) supplemented with L-glutamine and
gentamicin as above and 10% heat inactivated fetal calf serum
(Irvine Scientific) ~RPMI/10% FCS]. Chromium release assays
were performed in RPMI/10% FCS.

Cytokines. Recombinant human interleukin-2 (rIL-2) (Sandoz)
was used at a final concentration of 10 U/ml. Recombinant
human interleukin-7 (rIL-7) (Genzyme) was used at a final
concentration of 10 ng/ml.

Cultured Cell Lines. JY, a HLA A2.1 expressing human
EBV-transformed B-cell line, was grown in RPMI/10% FCS. K562,
a NK cell sensitive erythroblastoma line was grown in RPMI/10%

wo gS/~17 2 1 6 8 9 5 0 PCT~S94/08672

FCS. K562 was used to reduce background killing by NK and LAK
cells in the chromium release assays.

Peptides. The immunogenic peptides used in these studies were
synthesized as described above using motifs for HLA alleles
for specific target antigens as described in detail in
copending and commonly assigned applications U.S.S.N.
07/926,666 and U.S.S.N. 08/027,146 and their sequences are
shown in Table 1. Peptides were routinely dissolved in 100%
DMS0 at 20 mg/ml, aliquoted, and stored at -20C.

Isolation of Peripheral Blood Mononuclear Cells (PBMC). Whole
blood was collected in heparin (10 U/ml) containing syringes
and spun in 50cc conical centrifuge tubes (Falcon) at 1600 rpm
(Beckman GS-6KR) 15 min. The plasma layer was then removed
and 10 ml of the buffy coat collected with a 10 ml pipette
using a circular motion. The buffy coat was mixed thoroughly
and diluted with an equal volume of serum free RPMI 1640. The
diluted buffy coat was then layered over 20 ml Ficoll-Paque
(Pharmacia) in a 50cc conical tube and centrifuged 400xg for
20 minutes at room temperature without the brake off. The
interface containing the PBMCs was collected using a transfer
pipet (two interfaces per 50cc tube) and washed three times
with 50 ml serum free RPMI (1700, 1500, and 1300 rpm for 10
minutes.

Freezing and Thawing PBMC. PBMC were frozen at 30 x 106
cells/ml of 90% FCS + 10~ DMS0 (Sigma) in 1 ml aliquots using
cryovials (Nalge). Cryovials were placed in Cryo 1C freezing
containers (Nalge) containing isopropanol (Fisher) and placed
at -70C from 4 hours (minimum) to overnight (maximum).
Isopropanol was changed after every 5 uses. Cryovials were
transferred to liquid nitrogen for long term storage. PBMC
were thawed by continuous shaking in a 37C water bath until
the last crystal was nearly thawed. Cells were immediately
diluted into serum free RPMI medium containing DNAse 30 ~g/ml
(to avoid clumping by dead cells) (Calbiochem) and washed
twice.

W095/~17 21~ 8 9 5 0 PCT~S94/08672
21
Preparation of CD4+ T cell depleted responder cell population.
CD4+ lymphocyte depletion was performed using antibody-coated
flasks: MicroCELLector T-150 flasks for the selection of CD4+
cells (Applied Immune Sciences) were washed according to the
manufacturer's instructions with 25 ml PBS CMF (calcium
magnesium free) + 1 mM EDTA (Sigma) by swirling flasks for 30
sec followed by incubation for 1 hour at room temperature on a
flat surface. Buffer was aspirated and flasks were washed 2-
additional times by shaking the flasks for 30 seconds and
maintaining coverage of the binding surface. To each washed
flask, 25 ml culture medium were added and incubated for 20
minutes at room temperature on a flat surface. Media was left
in the flask until it was ready to receive the cells. PBMC
were thawed in culture medium containing 30 ~g/ml DNAse and
washed twice. For one flask a maximum of 12 x 107 cells were
resuspended in 25 ml culture medium. Culture medium was
aspirated from the flask and then the cell suspension was
gently added to the MicroC~TT~ctor. Flasks containing the
cells were incubated for 1 hour at room temperature on a flat
surface. At the end of the incubation, the flask was gently
rocked from side to side for 10 seconds to resuspend the
nonadherent cells. Nonadherent CD4+ T cell depleted cells
were harvested and then flasks were washed twice with PBS CMF
to collect the nonadherent cells. Harvested CD4+ T cell
depleted cells were pelleted by centrifugation and resuspended
in culture medium.

Generation of PHA Blasts. PBMC were isolated using the
stAn~Ard Ficoll-Paque protocol. Frozen cells were washed
twice before use. Cells were cultured at 2 x 106/ml in
RPMI/5% HS containing 1 ~g/ml PHA (Wellcome) and 10 U/ml
rIL-2. PHA blasts were maintained in culture medium
containing 10 U/ml rIL-2 with feeding and splitting as needed.
PHA blasts were used as APCs on day 6 of culture. Generation
of empty class I molecules and peptide loading was only
performed by the acid strip method when using PBMCs as APCs.

21 6 8 95 0 22 PCT~S94/08672
Acid Stripping/Peptide Loading of PBMC and PHA Blasts. PBMC
were isolated using the Ficoll-Paque protocol. When using
frozen cells, PBMC were washed twice before using. PHA blasts
were prepared as previously described and washed twice before
using. Once cells were prepared, they were washed once in
cold sterile 0.9% NaCl (J.T. Baker) + 1% BSA. In a 50cc
conical centrifuge tube, the cells were resuspended at 107/ml
in cold sterile citrate-phosphate buffer [0.13 M citric acid
(J.T. Baker), 0.06 M sodium phosphate monobasic (Sigma) pH 3,
1~ BSA, 3 ~g/ml ~2microglobulin (Scripps Labs)] and incubated
for 2 minutes on ice. Immediately, 5 volumes of cold sterile
neutralizing buffer #1 [0.15 M sodium phosphate monobasic pH
7.5, 1% BSA, 3 ~gtml ~2microglobulin, 10 ~g/ml peptide] were
added, and the cells were pelleted at 1500 rpm, 5 min at 4C.
Cells were resuspended in 1 volume cold sterile neutralizing
buffer #2 tPBS CMF, 1% BSA, 30 ~g/ml DNAse, 3 ~g/ml
microglobulin, 40 ~g/ml peptide] and incubated for 4 hours
at 20C. Cells were diluted with culture medium to
approximately 5 x 106/ml and irradiated with 6000 rads. Cells
were then centrifuged at 1500 rpm for 5 minutes at room
temperature and resuspended in culture medium. The acid
stripped/peptide loaded cells were used immediately in the CTL
induction cultures (below).

Binding Assays Using Intact Cells and Radiolabelled Peptide.
JY cells were either acid stripped (i.e. treated with
citrate-phosphate buffer and neutralizing buffer #1 as
described above) or incubated at a reduced temperature. JY
control cells were left untreated in tissue culture media.
After treatment both cell populations were washed twice with
serum free RPMI and loaded with 125I-radiolabelled 941.01 (HBc
18-27) peptide (st~n~rd chloramine T iodination). To
determine binding specificity, 2 x 106 cells were resuspended
in 200 ~l neutralizing buffer #2 (described above) containing
1251-941.01 (105 cpms) +/- 100 ~g unlabelled 941.01. Cells
were incubated for 4 hours at 20C and washed twice with serum
free RPMI to remove free peptide. Cells were resuspended in
200 ~l of serum free RPMI. In a microfuge tube the cell

W095/~17 2 16 8 ~ 5 ~ PCT~S94/08672

suspension was layered over an 800 ~l FCS and pelleted by
centrifugation for 5 seconds. Supernatants were aspirated and
the radioactivity remaining in the pellet was measured
(Micromedic automatic gamma counter, 1 minutes per tube).




Binding of Radiolabeled Peptides to Empty MHC Molecules. To
determine the efficiency of peptide loading using the cold
temperature incubation or acid stripping peptide loading
protocol, JY cells (an HLA-A2.1 EBV-transformed B cell line)
were preincubated at 26 C overnight or acid-stripped to remove
the endogenous MHC-associated peptides and the loading of
exogenous peptide was determined using a 125I-radiolabelled
HLA-A2.1 binding peptide. The specificity of this reaction
was determined by measuring the inhibition of labelled peptide
binding using a cold peptide of the same sequence. Results
presented in Table 2 demonstrate that acid-treatment of the
cells increased significantly (approximately 10-fold) the
amount of labelled peptide binding to the JY cells.
Furthermore, the binding of labelled peptide was completely
blocked by the addition of the cold peptide, demonstrating
specific binding (data not shown).

FACS Analysis. Approximately 106 cells were used for each
antibody that was to be tested. Cells were washed twice with
PBS CMF + 0.1% BSA. To each sample, 100 ~l PBS CMF + 0.1% BSA
+ primary antibody at 2 ~g/ml (BB7.2, ATCC) or (9.12.1,
INSERM-CNRS, Marseille) or (LB3.1, Children's Hospital,
Pittsburgh) were added. A negative control was always
included. Cells were incubated on ice for 20 minutes and
washed twice with PBS CMF + 0.1% BSA. Cells were resuspended
in 100 ~l anti-mouse IgG FITC conjugate (Sigma), diluted 1:50
in PBS CMF + 0.1% BSA, and incubated 20 minutes on ice. Cells
were washed twice with PBS CMF + 0.1% BSA, and resuspended in
PBS for FACScan (Becton Dickinson) analysis. When it was
necessary to postpone analysis to the subsequent days, the
cells were fixed with PBS/1% paraformaldehyde (Fisher) and
analyzed within one week.

WO95/~17 216 ~ 9 ~ O PCT~S94/08672
24
Measurements by FACS Analysis. PHA-induced T-cell blasts were
acid stripped/peptide loaded according to the methods
described above. The resulting cells were stained for FACS
analysis using anti-HLA-A2 (BB7.2) and anti-HLA alpha
chain-specific (9.12.1) monoclonal antibodies. Controls for
this experiment included the same cell population which was
not treated at pH 3 (but treated with PBS buffer at pH 7.2),
and cells treated with citrate-phosphate buffer (to strip the
MHC) but neutralized in the absence of ~2microglobulin and
peptide. The results presented in Figure 1, indicate ~hat
treatment of these cells with the citrate-phosphate (pH3)
buffer significantly reduced (10-fold) the reactivity of the
cells toward both anti-HLA class I antibodies alone
(anti-HLA-A2 and the alpha chain specific), but not towards a
monoclonal antibody specific for class II MHC molecules
(anti-HLA-DR). Most importantly, neutralization of the
acid-stripped cells in the presence of ~2microglobulin and
peptide resulted in preservation of a significant amount of
class I MHC antibody-reactive sites, with only a 2.5-fold
decrease in fluorescence intensity. The acid-treated cells
remained viable, as measured by trypan blue exclusion and
forward/lateral FACS scatter analysis. Similar results were
obtained using EBV-transformed B cell lines, fresh (or frozen)
PBMC and other peptides (which bind to either HLA-A2.1 or
HLA-Al) (data not shown).

Induction of Primary CTL using Acid Stripped/Peptide Loaded
Autologous PBMCs or PHA Blasts as Stimulators. Acid
stripping/peptide loading of PBMC and PHA blasts are described
above. During the 4 hour incubation of stimulator cells with
peptide, the responder cell population was prepared:
Responders were PBMC that were depleted of CD4+ T cells
(described above). Responder cells were resuspended in
culture medium at 3 x 106/ml and 1 ml of the responder cell
suspension was dispensed into each well of a 24-well tissue
culture plate (Falcon, Becton Dickinson). The plates were
placed in the incubator at 37C, 5% C02 until the stimulator
population was ready. Once irradiated, stimulator APCs were

wo 95~04817 ~ t 6 8 9 S O PCT/US94/08672


resuspended in culture medium containing 20 ng/ml rIL-7 at
106/ml for the PBMC, or at 3 x 105/ml for the PHA blasts, 1 ml
of stimulator cell suspension was added per well to the plates
containing the responders. On day 7 after induction, 100 ~l
5 culture medium containing 200 ng/ml rIL-7 was added to each
well (10 ng/ml rIL-7 final). On day 10 after induction, 100
~l of culture medium containing 200 U/ml rIL-2 was added to
each well (10 U/ml rIL-2 final).

10 Antigen Restimulation of CTL. On day 12-14 after the
induction, the primary CTL were restimulated with peptide
using autologous, adherent APCs. Autologous PBMC were thawed
and washed as described above. Cells were irradiated at 6000
rads. Cells were pelleted and resuspended in culture medium
at 4 x 106/ml and 1 ml of cell suspension was added to each
well of a 24-well tissue culture plate, and incubated for 2
hours at 37C, 5% CO2. Nonadherent cells were removed by
washing each well three times with serum free RPMI. After
this step, a 0.5 ml culture medium containing 3 ~g/ml
,l~2microglobulin and 20 ,ug/ml total peptide was added to each
well. APC were incubated for 2 hrs at 37C, under 5% CO2 with
the peptide and ~2microglobulin. Wells were aspirated and 1
ml of responder cells at 1.5 x 106/ml in culture medium was
added to each well. After 2 days, 1 ml of culture medium
containing 20 U/ml rIL-2 was added to each well. Cultures
were supplemented with 10 U/ml rIl-2 (final) every three days
thereafter.

Cytotoxicity Chromium Release Assay. Seven days following
restimulation of primary induction, the cytotoxic activity of
the cultures was assessed.
a. Effector Cell Preparation: The responders were
centrifuged and resuspended at 107/ml in RPMI/10% FCS.
Three-fold serial dilutions of effectors were performed to
yield effector to target ratios of 100:1, 33:1, 11:1, and 3:1.
Effector cells were aliquoted at 100 ~Ll/well on 96 well
U-bottomed cluster plates (Costar), in duplicate.

WO ~/~17 21 6 8 9 5 0 26 PCT~S~/08672

b. Target Cell PreParation: Approximately 16-20
hours prior to the assay, target cells were resuspended at 3 x
105/ml in RPMI/10% FCS in the presence or absence of 3 ~g/ml
~2microglobulin and 10 ~g/ml total peptide. After
preincubation, target cells were centrifuged and pellets were
resuspended in 200 ~1 (300~Ci) sodium (51Cr) chromate (NEN).
Cells were incubated at 37C for 1 hour with agitation.
Labelled target cells were washed 3 times with RPMI/10% FCS.
c. Settinq-Up the Assays: Target cell
concentration was adjusted to 105/ml in RPMI/10% FCS and 100
~1 aliquots were added to each well containing responders.
K562 cells (cold targets, to block NK, and LAK activity) were
washed and resuspended in RPMI/10% FCS at 107/ml. Aliquots of
20 ~1 were added per well, yielding a 20:1 cold K562 target to
labelled target ratio. For the determination of the
spontaneous 51Cr release, 100 ~l/well of RPMI/10% FCS were
added to 100 ~l/well of labelled target cells, and 20 ~l/well
of K562. For maximum 51Cr release, 100 ~1 1% Triton X-100
(Sigma) in PBS CMF, was added to the 100 ~l/well labelled
target cells, and 20 ~l/well K562. Plates were centrifuged
for 2 minutes at 1200 rpm to accelerate cell conjugate
formation. Assays were incubated for 5 hours at 37C, 5~ C02.
Assays were harvested by centrifuging plates for 5 minutes at
1200 rpm and collecting 100 ~l/well of supernatant. St~n~Ard
gamma counting techniques were used to determine percent
specific lysis (Micromedic automatic gamma counter, 0.5
minutes per tube). Percent specific lysis was determined by
the following formula: cpm experimental release -cpm
spontaneous release/cpm maximum release-cpm spontaneous
release x 100.

In Vitro Induction of Primary Antigen-Specific CTL Using Acid
Stripped/Peptide Loaded APCS. Additional critical parameters
for the induction of primary CTL are: 1) enrichment of CD8+
T-cells in the responder cell population (by depletion of CD4+
T-cells), 2) addition of rIL-7 to the CTL induction cultures
from day 0, and 3) restimulation of the cultures with antigen
on day 12-14 using autologous adherent cells pulsed with

wo 95/04817 2 I S ~ 9 ~ ~ PCT~Sg4/08672

peptide. Results presented in Figures 2, 3 and 5 correspond
to experiments performed using PBMC as APC. The results
presented in Figure 4 present results obtained using
PHA-induced T-cell blasts as APC. Figure 7 shows a comparison
of the acid strip loading technique (Figure 7a) to the cold
temperature incubation technique (Figure 7b).

Example 3
Screeninq Peptides to identify CTL epitopes.
In order to identify CTL epitopes, CTL were
stimulated by SAC-I activated PBMCs as APC. Cold temperature
enhanced expression of empty MHC enabling loading of antigenic
peptide to generate SAC-I activated PBMC APC. This method
presents an alternative protocol to the methods described
above for the generation of the APC which are used to
stimulate CTL. This example also presents an alternative
protocol for the stimulation of CTL by the APC.

Complete Culture Medium. The tissue culture medium used in
this study consisted of RPMI 1640 with Hepes and L-glutamine
(Gibco) (Biowhittaker) supplemented with 2 mM L-glutamine
(Irvine Scientific), 0.5mM sodium pyruvate (Gibco), 100 U/100
ug/ml penicillin/streptomycin (Irvine), and 5%
heat-inactivated Human Serum Type AB (RPMI/5% HS; Gemini
Bioproducts). Culture media used in the growth of
EBV-transformed lines contained 10% heat-inactivated fetal
calf serum (RPMI/10% FCS, Irvine) instead of human serum.

Cytokines. Recombinant human Interleukin-2 (rIL-2) and
Interleukin-4 (rIL-4) were obtained from Sandoz and used at a
final concentration of 10 U/ml and 10 ng/ml, respectively.
Human interferon-~ (IFN-~) and recombinant human Interleukin-7
(rlL-7) were obtained from Genzyme and used at 20 U/ml and 10
ng/ml, respectively.

Peptides. Peptides were synthesized as described above and
are described in Table 1. Peptides were routinely dissolved

WO95/~17 ~1 6 8 9 ~ o PCT~S94108672
28
in 100~ DMSO at 20 mg/ml, aliquoted, and stored at -70C until
used.

Cell Lines. JY, Steinlein, EHM, BVR, and KT3 are homozygous
S human EBV-transformed B cell lines expressing HLA A2 1, A1, A3,
A11, and A24, respectively. They are grown in RPMI/10% FCS
and used as targets in the CTL assays. K562, an NK cell
sensitive, erythroblastoma line grown in RPMI/10% FCS, was -
used for reduction of background killing in the CTL assays.
Melanoma HLA A1+ cell lines either expressing the MAGE
antigen, mel 397 and mel 938 or those not expressing the MAGE
antigen, mel 888, were also grown in RPMI/10% FCS.

Isolation of Peripheral Blood Mononuclear Cells (PBMCs).
Whole blood was collected into heparin containing syringes and
spun in 50cc tubes at 1600 RPM (Beckman GS-6KR) for 15
minutes. The plasma layer was then removed and 10 ml of buffy
coat was collected with a pipette using a circular motion.
The buffy coat was mixed well and diluted with an equal volume
of RPMI. The buffy coat (30 ml) was then layered on 20 ml of
Ficoll-Paque (Pharmacia) and centrifuged at 1850 RPM (400xg)
for 20 minutes, 25C, with the brake off. The interface
between the Ficoll and the plasma containing the PBMCs was
recovered with a transfer pipet (two interfaces per 50 ml
tube) and washed three times with 50 ml of RPMI (1700, 1500,
and 1300 RPM for 10 minutes). Cells were resuspended in 10-20
ml of culture medium, counted, and adjusted to the appropriate
concentration.

Freezing PBMCs. 30 million cells/tube (90% FCS/10% DMSO;
Sigma) were inserted into a Nalgene Cryo 1C Freezing
Container containing isopropanol (Fisher) and placed at -70C
from 4 hrs (minimum) to overnight (maximum). The isopropanol
was changed every five times. Tubes were transferred to
liquid nitrogen for long term storage. To thaw, PBMCs were
continuously shaken in a 37C water bath until the last
crystal was almost thawed (tubes were not allowed to sit in
the water bath or at room temperature for any period of time).

wo gS/~17 2 ~ 6 ~ 9 ~ ~ PCT~S94/08672
29
Cells were diluted into serum-free RPMI containing 30 ~g/ml
DNase to prevent clumping by dead cell DNA and washed twice.

Induction of Primary CTL Using SAC-I Activated PBMCs as APCs
a. PreParation of SAC-I activated PBMCs as APCs: PBMCs
were purified using the standard Ficoll-Paque protocol and
resuspended at 1 x 106/ml in RPMI/5% FCS containing 0.005%
Pansorbin cells (SAC-I cells expressing Protein A;
Calbiochem), 20 ~g/ml Immunobeads (Rabbit anti-Human IgM;
Biorad), and 20 ng/ml of human rIL-4. Two ml of cells per
well were plated in a 24-well plate (Falcon, Becton Dickinson)
and cultured at 37C. After 3 days, the medium was removed
and the cells were washed three times followed by addition of
RPMI/10% HS. The cells were used after culturing for an
additional 2 days in RPMI/10% HS.

b. Ex~ression of emptY Class I molecules on the surface
of APCs and Peptide loading of APCs.
1. Cold temperature incubation:
a. Expression of empty MHC in APCs: The APCs
were adjusted to a concentration of 2 x 106/ml in complete
culture medium containing 10 ng/ml rIL-4, 20 U/ml human IFN-~,
and 3 ~g/ml ~2-microglobulin (~2m; Scripps Lab). The cells
were then incubated overnight at 26C in the presence of 5%
CO2. It should be noted that these cells only express a
fraction of Class I molecules in the empty state (-10%).
b. Peptide loading of APC stimulator cells:
Empty Class I expressing APCs were washed 1-2 times with
serum free RPMI (+ L-glutamine and Hepes) and resuspended at 1
X 107 in serum-free RPMI containing 50 ~g/ml total of the
peptide pool (i.e., 16.7 ~g/ml of each peptide in a pool of
three; 25 ~g/ml of each peptide in a pool of two; 50 ~g/ml of
individual peptide), 30 ~g/ml DNAse, and 3 ~g/ml ~2m.
Following a 4 hour incubation at 20C, the cells were
irradiated at 6100 rads (5 x 106/ ml; 25 million cells/tube),
washed and adjusted to the appropriate concentration for
addition to the induction culture (see below).
2. Acid stripping: This was used as an

WO95/~17 216 8 9 ~ ~ PCT~S94/08672

alternative method for generating empty MHC on the surface of
the APCs. The SAC-I activated PBMCs were washed once in cold
0.9% sodium chloride (J.T. Baker) containing 1% BSA. The
cells were resuspended at 107/ml in cold citrate-phosphate
buffer (0.13M citric acid tJ.T. Baker], 0.06M sodium phosphate
monobasic [Sigma], pH3) containing 1% BSA and 3 ~g/ml ~2m and
incubated on ice. After 2 minutes, 5 volumes of cold 0.15M
sodium phosphate buffer, pH7.5, containing 1% BSA, 3 ~g/ml -
~2m, and 10 ~g/ml peptide [neutralizing buffer #1] was added
and the cells centrifuged at 1500 RPM for 5 minutes at 4C.
The cells were resuspended in 1 ml of cold PBS containing 1
BSA, 30 ~g/ml DNase, 3 ~g/ml ~2microglobulin, and 50 ~g/ml
peptide [neutralizing buffer #2] and incubated for 4 hours at
20C. As above, subsequent to the four hour incubation at
20C, the cells were irradiated at 6100 rads (5 x 106/ ml; 25
million cells/tube), washed, then adjusted to the appropriate
concentration for addition to the induction culture (see
below).

c. Prearation of the CD4+ depleted PBMC resPonder cell
population (depletion of lYmphocvte subPoPulations usinq
AIS flasks). AIS MicroCellector T-150 flasks (specific
for the depletion of CD4+ T cells; Menlo Park, CA) were
primed by adding 25 ml of PBS/1 mM EDTA, swirling for 30
seconds so that all surfaces were moistened, and then
incubating with the binding surface down at room temperature
for 1 hour. Following this incubation, flasks were shaken
vigorously for 30 seconds, washed 1 time with PBS/EDTA, 2
additional times with PBS and then incubated with 25 ml of
culture medium for 15 minutes. PBMCs were thawed in
serum-free RPMI (+ L-glutamine + Hepes) containing 30 ~g/ml
DNAse, washed once, and incubated for 15 minutes in culture
medium. Following aspiration of culture medium from the
flasks, up to 180 million PBMCs were added in 25 ml of culture
medium containing 30 ~g/ml DNAse. After 1 hour at room
temperature, the flasks were rocked gently for 10 seconds to
resuspend the nonadherent cells. The nonadherent cell
suspension containing the CD8+ T cells was collected and the

WO951~17 ~1 6 8 9 ~ O PCT~S94/08672

flasks were washed 2 times with PBS. The CD4+ T cell depleted
PBMCs were centrifuged and counted for addition to the
induction culture. The CD4+ and CD8+ phenotype of the CD4+
depleted cell population was determined by FACS analysis (see
below). In general, this technique resulted in a two-fold
enrichment for CD8+ T cells with an average of approximately
40-50% CD8+ T cells and 15-20% remaining CD4+ T cells
following depletion of CD4+ T cells. Depletion of CD4+ T
cells can also be accomplished by using antibody and
complement methods or antibody coated magnetic beads
(Dynabeads). Depletion of CD4+ T cells enriched the CTLp and
removed cells which competed for cell nutrients.
d. Induction of primarY CTL. During the 4 hour peptide
loading of the stimulator APCs, CD4+ depleted PBMC to be used
as the responder population were prepared utilizing AIS flasks
for selection of CD8+ T cells through the depletion of CD4+ T
cells (above). The responder cells were plated at 3 x 106/ml
in a 1 ml volume (24 well plate) and placed at 37C until the
peptide loaded stimulator APCs were prepared. The irradiated,
peptide loaded APCs were washed l time in serum-free RPMI (+
L-glutamine and Hepes), adjusted to the appropriate
concentration in complete medium, and plated into a 24 well
plate at 1 ml/plate: For PBMC and SAC-I activated PBMCs as
APCs 1 x 106 stimulator cells (1 ml volume) were plated into
the wells containing the responder cells; For PHA blasts as
APCs, 1 ml of 3 x 105/ml stimulator cells were plated in each
well. A final concentration of 10 ng/ml of rIL-7 (2 ml total
volume) was added. On day 7 an additional 10 ~g/ml rIL-7 was
added to the culture and 10 U/ml rIL-2 was added every 3 days
thereafter. On day 12, the cultures were restimulated with
peptide pulsed adherent cells and tested for cytolytic
activity 7 days later (below).

Protocol for Restimulation of Primary CTL Using Autologous
Adherent APC. Autologous PBMCs were thawed into serum-free
RPMI (+ L-glutamine and Hepes) containing 30~g/ml DNAse,
washed 2 times, and adjusted to 5 x 106 /ml in culture medium
containing DNAse. PBMCs (25 million cells/tube in 5 ml) were

WO95/~17 ~ ~ 8 9 5 o PCT~S94/08672

32
irradiated at 6100R. After 1 wash, the PBMCs were resuspended
in culture medium and adjusted to 4 x 106/ml and 1 ml of
irradiated PBMCs was added per well of a 24-well plate. The
PBMC were incubated for 2 hours at 37C, washed 3 times to
remove nonadherent cells, and cultured in medium containing 20
~g/ml total peptide and 3 ~g/ml ~2microglobulin added in a 0.5
ml volume and again incubated for 2 hours at 37C. The
peptide was aspirated and 1.5 x 106 responder cells
resuspended in culture medium were added in a 1 ml volume.
After 2 days, 1 ml of culture medium containing 20 U/ml rIL-2
was added.
FACS Analysis. One million cells/tube were centrifuged,
resuspended in 100 ~l/tube PBS/0.1~ BSA/0.02% sodium azide
(Sigma) plus 10 ~l/tube directly conjugated antibody (Becton
Dickinson), and incubated on ice 15-20 minutes. Cells were
then washed 2 times with PBS/0.1% BSA/0.02% sodium azide and
resuspended in PBS to analyze on FACScan (Becton Dickinson).
When it was not possible to analyze samples within 1-2 days,
cells were fixed with PBS containing 1% paraformaldehyde
(Fisher) and analyzed within one week.

Cytotoxicity Assay
a. Tarqet cell ~re~aration. Approximately 16-20 hours
prior to the CTL assay, target cells (Class I matched
EBV-transformed lines) were washed once and resuspended in a
10 ml volume at 3 x 105/ml in RPMI/5% FCS in the presence or
absence of 10 ~g/ml total peptide.
b. Labeling of tarqet cells: Target cells were
centrifuged and resuspended in 200 ~l/tube sodium 51Cr
chromate (NEN), then incubated at 37C for 1 hour on a shaker.
Targets were washed 3 times (10 ml/wash) with RPMI/10% FCS and
resuspended in 10 ml (to determine the efficiency of
labelling, 50 ~l/target was counted on the Micromedic
automatic gamma counter).
c. CTL assay. Target cells were adjusted to 2 x 105/ml
and 50 ~1 of the cell culture was added to each well of a
U-bottomed 96-well plate (Costar Corp.) for a final
concentration of 1 X 104/well. K562 cells were washed once,

WO95/~17 PCT~S94/08672
~ 1 689~0
33
resuspended at 4 x 106/ml, and 50 ~l/well was added for a
final concentration of 2 x 105/well (ratio of cold K562 to
target was 20:1). Responder cells were washed once,
resuspended at 9 x 106/ml, and three fold serial dilutions
were performed for effector to target ratios of 90:1, 30:1,
10:1, and 3:1. Responder cells were added in a volume of 100
~l in duplicate wells. For spontaneous release, 50 ~l/well of
labelled target cells, 50 ~l/well K562, and lOo ~l/well of
medium was added. For maximum release, 50 ~l/well target, 50
~l/well K562, and 100 ~l/well of 0.1% Triton-X100 (Sigma) was
added. Plates were centrifuged for 5 minutes at 1200 RPM.
Following a 5 hour incubation at 37OC, plates were centrifuged
again for 5 minutes at 1200 RPM, and lOo ~l/well of
supernatant was collected. Standard gamma counting techniques
(Micromedic automatic gamma counter; 0.5 minutes/tube) were
used to determine the percent specific lysis according to the
formula: % specific lysis = cpm experimental release - cpm
spontaneous release/cpm maximum release - cpm spontaneous
release X 100. A cytotoxicity assay (CTL assay) was
considered positive if the lysis by CTL of targets sensitized
with a specific peptide at the two highest effector to target
(E:T) ratios was 15% greater than lysis of control targets
(i.e. target cells without peptide). A cytotoxicity assay
(CTL assay) was considered borderline if the lysis by CTL of
targets sensitized with a specific peptide at the two highest
effector to target (E:T) ratios was 6% greater than lysis of
control targets (i.e. target cells without peptide).
d. Results. of the peptides that bind to the indicated
alleles, 12 of the 60 MAGE peptides, 13 of the 53 HIV
peptides, 3 of the 25 HCV peptides, and 7 of the 28 HBV
peptides tested to date induced primary CTL in vitro.
Representative graphs illustrating CTL responses to various
immunogenic peptides are shown for MAGE (Figure 8), HIV
(Figure 9), HCV (Figure 10), and HBV (Figure 11). The CTL
induction data is summarized in Table 3 which lists the
immunogenic peptides which bind to the appropriate MHC and
induce primary CTL in vitro. Indicated is the peptide's
sequence, corresponding antigen and HLA allele to which it

WO95/04817 ~1 6 ~ 9 ~ 9 PCT~S94/08672
34
binds. Results shown in Figure 6 illustrate lysis of peptide
sensitized targets and endogenous targets following
stimulation with SACI activated PBMCs loaded with the
immunogenic peptide MAGE-3 1044.07 which had been loaded using
cold temperature incubation.

Although the present invention has been described in
some detail by way of illustration and example for purposes-of
clarity and understanding, it will be apparent that certain
changes and modifications may be practiced within the scope of
the appended claims.

WO 95/04817 PCT/US94/08672
216$~

Table 1


- Peptides Synthesized For Loading Onto Acid Stripped Autologous
PBMCs and PHA Blasts
Peptide ID # Antigen Sequence
777.03 HBVs 20-28 FLLTRILTI
924.07 HBVc 18-27 FLPSDFFPSV
927.32 HBVp 61-69 GLYSSTVPV
938.01 MAGE 1 161-169 EADPTGHSY
939.03 PSA 49-57 VLVHPQWVL
941.01 HBVc 18-27 analog FLPSDYFPSV
1044.04 PAP 135-143 ILLWDPIPV
1044.05 PSA 166-175 KLQCVDLVHI -
1044.06 PSA 118-128 MLLRLSEPAEL
1044.07 MAGE 3 161-169 EVDPIGHLY
1044.01 MAGE 3 8-17 ASSLY~
1072.13 MAGE 1 96-104 SLFRAVITK
1072.18 MAGE 1 66-74 TTINFTRQR
1072.20 MAGE 1 219-227 SVMEVYDGR
1072.39 MAGE lN 270-279 RALAETSYVK
1072.22 MAGE 1 238-247 LLTQDLVQEK
1072.15 MAGE 1 95-104 ESLFRAVITK
1069.42 HIV pol 1225-1235 KVYLAWVPAHK
1069.43 HIV env 2185-2194 TVYYGVPVWK
1069.46 HIV pol 1032-1042 WTYQIYQEPFK

wo 95~817 21 6835 o 36 PCT~S94/08672


1069.47 HIV env 2184-2194 VTVYYGVPVWK
1069.48 HIV pol 1434-1443 AVFIHNFKRK
1073.10 HCV LORF 1858-1867 GVAGALVAFK
1073.11 HCV CORE 43-51 RLGVRATRK
1073.16 HCV LORF 1227-1236 HLHAPTGSGK
1069.15 HBV pol 724-733 TLWKAGILYK

WO95/04817 PCT~S94/08672

Table 2
Acid Stripping, Peptide Loading of JY Cells with Radiolabelled
941.01
Cell Population 125I-Labeled CPMS
Peptide +/- Cold +/- std.
Peptide dev.
JY acid stripped - cold peptide 3553 + 157 n = 3
JY acid stripped + cold peptide 13 n = 1
JY control -cold peptide 370 + 37 n = 3
JY control + cold peptide 50 n = 1

WO 95/04817 216 8 9 38 PCT/US94/08672



Table 3




8~ ~c- Adi~ Molif Id
EVDPIt;~LY M~GE3 A01 1044.07
ASSLP ~ MAGE3 A01 1044.0l
EADPTGHSY MAGEI A01 9S8.01
SSI.PT~Y~ MAGE3 A01 1072.02
GSWtiNWQY~ M~GE3 ~01 1072.03
AL~ETSYVX~ MAGElN A03 1072.38
SLFR~VIT~ MAGEl Al I lm2.13
RAL1~ETSYV~CMAGElN Al 1 1072.39
ESLFRAVITX MAGEl ~11 1072.1S
xv~rLAmA~ ~v ~3/ll~ 1069.42
Y~,VPVWX ~ 03 .069.43
WPVK HIV A03 .069.44
GGI~r~lJ~ ~V ~05 069.4S
AIFQSSM'rX ~IIV A0., 966Ø
~ISYQ~ IV A0~ 6
FLGRIWPS~ IIV kO~ l069.56
r~NX ~V Al 10S' 03
,vr~ IIV Al l 106'~.47
GVAGaLVAFK HCV A03 lO73.10
,GSSDI.Y HCV A11 10~9.62
GVAGALVAFK HCV A11 lO~ OS
LLDTASALY ~ H~V AOI ~1069.01
~LWXAGILYX HBV ~03 1069.1S
bord~rllne ~osl~e

Representative Drawing

Sorry, the representative drawing for patent document number 2168950 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 1994-08-01
(87) PCT Publication Date 1995-02-16
(85) National Entry 1996-02-06
Examination Requested 2001-11-14
Dead Application 2005-08-01

Abandonment History

Abandonment Date Reason Reinstatement Date
1999-08-03 FAILURE TO PAY APPLICATION MAINTENANCE FEE 2000-01-31
2001-08-01 FAILURE TO REQUEST EXAMINATION 2001-11-14
2004-08-02 FAILURE TO PAY APPLICATION MAINTENANCE FEE
2004-09-02 R30(2) - Failure to Respond
2004-09-02 R29 - Failure to Respond

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $0.00 1996-02-06
Maintenance Fee - Application - New Act 2 1996-08-01 $100.00 1996-07-16
Registration of a document - section 124 $0.00 1996-08-01
Registration of a document - section 124 $50.00 1997-03-04
Maintenance Fee - Application - New Act 3 1997-08-01 $100.00 1997-08-01
Maintenance Fee - Application - New Act 4 1998-08-03 $100.00 1998-07-16
Reinstatement: Failure to Pay Application Maintenance Fees $200.00 2000-01-31
Maintenance Fee - Application - New Act 5 1999-08-03 $150.00 2000-01-31
Maintenance Fee - Application - New Act 6 2000-08-01 $150.00 2000-07-21
Maintenance Fee - Application - New Act 7 2001-08-01 $150.00 2001-06-26
Reinstatement - failure to request examination $200.00 2001-11-14
Request for Examination $200.00 2001-11-14
Back Payment of Fees $200.00 2001-11-14
Maintenance Fee - Application - New Act 8 2002-08-01 $150.00 2002-07-19
Maintenance Fee - Application - New Act 9 2003-08-01 $150.00 2003-07-23
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
EPIMMUNE INC.
Past Owners on Record
CELIS, ESTEBAN
CYTEL CORPORATION
KUBO, RALPH
SERRA, HORACIO
TSAI, VAN
WENTWORTH, PEGGY
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Claims 1995-02-16 3 90
Drawings 1995-02-16 10 144
Description 1995-02-16 38 1,864
Cover Page 1996-05-30 1 19
Abstract 1995-02-16 1 41
Assignment 1996-02-06 18 678
PCT 1996-02-06 10 486
Prosecution-Amendment 2001-06-15 1 46
Prosecution-Amendment 2001-11-14 1 45
Prosecution-Amendment 2004-03-02 3 93